Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 951
Filter
1.
J Cell Biol ; 223(12)2024 Dec 02.
Article in English | MEDLINE | ID: mdl-39352499

ABSTRACT

Successful axonal regeneration following injury requires the effective allocation of energy. How axons withstand the initial disruption in mitochondrial energy production caused by the injury and subsequently initiate regrowth is poorly understood. Transcriptomic data showed increased expression of glycolytic genes after optic nerve crush in retinal ganglion cells with the co-deletion of Pten and Socs3. Using retinal cultures in a multicompartment microfluidic device, we observed increased regrowth and enhanced mitochondrial trafficking in the axons of Pten and Socs3 co-deleted neurons. While wild-type axons relied on mitochondrial metabolism, after injury, in the absence of Pten and Socs3, energy production was supported by local glycolysis. Specific inhibition of lactate production hindered injury survival and the initiation of regrowth while slowing down glycolysis upstream impaired regrowth initiation, axonal elongation, and energy production. Together, these observations reveal that glycolytic ATP, combined with sustained mitochondrial transport, is essential for injury-induced axonal regrowth, providing new insights into the metabolic underpinnings of axonal regeneration.


Subject(s)
Axons , Glycolysis , Mitochondria , Nerve Regeneration , Retinal Ganglion Cells , Animals , Axons/metabolism , Nerve Regeneration/genetics , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Mitochondria/metabolism , Mitochondria/genetics , Mice , Optic Nerve Injuries/metabolism , Optic Nerve Injuries/pathology , Optic Nerve Injuries/genetics , PTEN Phosphohydrolase/metabolism , PTEN Phosphohydrolase/genetics , Mice, Inbred C57BL , Adenosine Triphosphate/metabolism , Energy Metabolism/genetics
2.
BMC Genomics ; 25(1): 896, 2024 Sep 29.
Article in English | MEDLINE | ID: mdl-39343885

ABSTRACT

BACKGROUND: Decellularized extracellular matrix (dECM) is an intriguing natural biomaterial that has garnered significant attention due to its remarkable biological properties. In our study, we employed a cell-matrixed nerve graft for the repair of sciatic nerve defects in rats. The efficacy of this approach was assessed, and concurrently, the underlying molecular regulatory mechanisms were explored to elucidate how such grafts facilitate nerve regeneration. Long noncoding RNAs (lncRNAs) regulate mRNA expression via multiple mechanisms, including post-transcriptional regulation, transcription factor effects, and competitive binding with miRNAs. These interactions between lncRNAs and mRNAs facilitate precise control of gene expression, allowing organisms to adapt to varying biological environments and physiological states. By investigating the expression profiles and interaction dynamics of mRNAs and lncRNAs, we can enhance our understanding of the molecular mechanisms through which cell-matrixed nerve grafts influence neural repair. Such studies are pivotal in uncovering the intricate networks of gene regulation that underpin this process. RESULTS: Weighted gene co-expression network analysis (WGCNA) utilizes clustering algorithms, such as hierarchical clustering, to aggregate genes with similar expression profiles into modules. These modules, which potentially correspond to distinct biological functions or processes, can subsequently be analyzed for their association with external sample traits. By correlating gene modules with specific conditions, such as disease states or responses to treatments, WGCNA enables a deeper understanding of the genetic architecture underlying various phenotypic traits and their functional implications. We identified seven mRNA modules and five lncRNA modules that exhibited associations with treatment or time-related events by WGCNA. We found the blue (mRNAs) module which displayed a remarkable enrichment in "axonal guidance" and "metabolic pathways", exhibited strong co-expression with multiple lncRNA modules, including blue (related to "GnRH secretion" and "pyrimidine metabolism"), green (related to "arginine and proline metabolism"), black (related to "nitrogen metabolism"), grey60 (related to "PPAR signaling pathway"), and greenyellow (related to "steroid hormone biosynthesis"). All of the top 50 mRNAs and lncRNAs exhibiting the strongest correlation were derived from the blue module. Validation of key molecules were performed using immunohistochemistry and qRT-PCR. CONCLUSION: Revealing the principles and molecular regulatory mechanisms of the interaction between materials and biological entities, such as cells and tissues, is a direction for the development of biomimetic tissue engineering technologies and clinically effective products.


Subject(s)
Nerve Regeneration , RNA, Long Noncoding , RNA, Messenger , Sciatic Nerve , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Animals , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Nerve Regeneration/genetics , Sciatic Nerve/injuries , Sciatic Nerve/metabolism , Extracellular Matrix/metabolism , Gene Regulatory Networks , Gene Expression Profiling , Rats, Sprague-Dawley
3.
PLoS One ; 19(9): e0304773, 2024.
Article in English | MEDLINE | ID: mdl-39231134

ABSTRACT

Application of polyethylene glycol (PEG) to a peripheral nerve injury at the time of primary neurorrhaphy is thought to prevent Wallerian degeneration via direct axolemma fusion. The molecular mechanisms of nerve fusion and recovery are unclear. Our study tested the hypothesis that PEG alters gene expression in neural and muscular environments as part of its restorative properties. Lewis rats underwent unilateral sciatic nerve transection with immediate primary repair. Subjects were randomly assigned to receive either PEG treatment or standard repair at the time of neurorrhaphy. Samples of sciatic nerve distal to the injury and tibialis muscle at the site of innervation were harvested at 24 hours and 4 weeks postoperatively. Total RNA sequencing and subsequent bioinformatics analyses were used to identify significant differences in differentially expressed genes (DEGs) and their related biological pathways (p<0.05) in PEG-treated subjects compared to non-PEG controls. No significant DEGs were identified in PEG-treated sciatic nerve compared to controls after 24 hours, but 1,480 DEGs were identified in PEG-treated tibialis compared to controls. At 4 weeks, 918 DEGs were identified in PEG-treated sciatic nerve, whereas only 3 DEGs remained in PEG-treated tibialis compared to controls. DEGs in sciatic were mostly upregulated (79%) and enriched in pathways present during nervous system development and growth, whereas DEGs in muscle were mostly downregulated (77%) and related to inflammation and tissue repair. Our findings indicate that PEG application during primary neurorrhaphy leads to significant differential gene regulation in the neural and muscular environment that is associated with improved functional recovery in animals treated with PEG compared to sham non-PEG controls. A detailed understanding of key molecules underlying PEG function in recovery after peripheral nerve repair may facilitate amplification of PEG effects through systemic or focal treatments at the time of neurotmesis.


Subject(s)
Muscle, Skeletal , Peripheral Nerve Injuries , Polyethylene Glycols , Rats, Inbred Lew , Sciatic Nerve , Animals , Rats , Sciatic Nerve/injuries , Peripheral Nerve Injuries/genetics , Polyethylene Glycols/pharmacology , Muscle, Skeletal/metabolism , Muscle, Skeletal/innervation , Muscle, Skeletal/drug effects , Disease Models, Animal , Sequence Analysis, RNA , Nerve Regeneration/drug effects , Nerve Regeneration/genetics , Male , Gene Expression Regulation/drug effects , Gene Expression Profiling
4.
Proc Natl Acad Sci U S A ; 121(38): e2402518121, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39254997

ABSTRACT

The in vivo three-dimensional genomic architecture of adult mature neurons at homeostasis and after medically relevant perturbations such as axonal injury remains elusive. Here, we address this knowledge gap by mapping the three-dimensional chromatin architecture and gene expression program at homeostasis and after sciatic nerve injury in wild-type and cohesin-deficient mouse sensory dorsal root ganglia neurons via combinatorial Hi-C, promoter-capture Hi-C, CUT&Tag for H3K27ac and RNA-seq. We find that genes involved in axonal regeneration form long-range, complex chromatin loops, and that cohesin is required for the full induction of the regenerative transcriptional program. Importantly, loss of cohesin results in disruption of chromatin architecture and severely impaired nerve regeneration. Complex enhancer-promoter loops are also enriched in the human fetal cortical plate, where the axonal growth potential is highest, and are lost in mature adult neurons. Together, these data provide an original three-dimensional chromatin map of adult sensory neurons in vivo and demonstrate a role for cohesin-dependent long-range promoter interactions in nerve regeneration.


Subject(s)
Axons , Chromatin , Cohesins , Nerve Regeneration , Promoter Regions, Genetic , Sensory Receptor Cells , Animals , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/physiology , Mice , Promoter Regions, Genetic/genetics , Chromatin/metabolism , Nerve Regeneration/genetics , Nerve Regeneration/physiology , Axons/metabolism , Axons/physiology , Humans , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Enhancer Elements, Genetic/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Ganglia, Spinal/metabolism , Ganglia, Spinal/cytology , Sciatic Nerve/metabolism
5.
PLoS Genet ; 20(8): e1011388, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39186815

ABSTRACT

Most neurons are not replaced after injury and thus possess robust intrinsic mechanisms for repair after damage. Axon injury triggers a calcium wave, and calcium and cAMP can augment axon regeneration. In comparison to axon regeneration, dendrite regeneration is poorly understood. To test whether calcium and cAMP might also be involved in dendrite injury signaling, we tracked the responses of Drosophila dendritic arborization neurons to laser severing of axons and dendrites. We found that calcium and subsequently cAMP accumulate in the cell body after both dendrite and axon injury. Two voltage-gated calcium channels (VGCCs), L-Type and T-Type, are required for the calcium influx in response to dendrite injury and play a role in rapid initiation of dendrite regeneration. The AC8 family adenylyl cyclase, Ac78C, is required for cAMP production after dendrite injury and timely initiation of regeneration. Injury-induced cAMP production is sensitive to VGCC reduction, placing calcium upstream of cAMP generation. We propose that two VGCCs initiate global calcium influx in response to dendrite injury followed by production of cAMP by Ac78C. This signaling pathway promotes timely initiation of dendrite regrowth several hours after dendrite damage.


Subject(s)
Adenylyl Cyclases , Calcium Channels, L-Type , Calcium , Cyclic AMP , Dendrites , Animals , Adenylyl Cyclases/metabolism , Adenylyl Cyclases/genetics , Axons/metabolism , Axons/physiology , Calcium/metabolism , Calcium Channels/metabolism , Calcium Channels/genetics , Calcium Channels, L-Type/metabolism , Calcium Channels, L-Type/genetics , Calcium Channels, T-Type/metabolism , Calcium Channels, T-Type/genetics , Calcium Signaling/genetics , Cyclic AMP/metabolism , Dendrites/metabolism , Drosophila/genetics , Drosophila melanogaster/genetics , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Nerve Regeneration/physiology , Nerve Regeneration/genetics , Neurons/metabolism , Regeneration/genetics , Regeneration/physiology , Signal Transduction
6.
Int J Mol Sci ; 25(16)2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39201743

ABSTRACT

Neurodegenerative disorders, including traumatic injuries to the central nervous system (CNS) and neurodegenerative diseases, are characterized by early axonal damage, which does not regenerate in the adult mammalian CNS, leading to permanent neurological deficits. One of the primary causes of the loss of regenerative ability is thought to be a developmental decline in neurons' intrinsic capability for axon growth. Different molecules are involved in the developmental loss of the ability for axon regeneration, including many transcription factors. However, the function of microRNAs (miRNAs), which are also modulators of gene expression, in axon re-growth is still unclear. Among the various miRNAs recently identified with roles in the CNS, miR-17, which is highly expressed during early development, emerges as a promising target to promote axon regeneration. Here, we used adeno-associated viral (AAV) vectors to overexpress miR-17 (AAV.miR-17) in primary cortical neurons and evaluate its effects on neurite and axon regeneration in vitro. Although AAV.miR-17 had no significant effect on neurite outgrowth and arborization, it significantly enhances neurite regeneration after scratch lesion and axon regeneration after axotomy of neurons cultured in microfluidic chambers. Target prediction and functional annotation analyses suggest that miR-17 regulates gene expression associated with autophagy and cell metabolism. Our findings suggest that miR-17 promotes regenerative response and thus could mitigate neurodegenerative effects.


Subject(s)
Axons , Dependovirus , MicroRNAs , Nerve Regeneration , Neurites , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Axons/metabolism , Axons/physiology , Nerve Regeneration/genetics , Neurites/metabolism , Dependovirus/genetics , Cells, Cultured , Genetic Vectors/genetics , Mice , Neurons/metabolism
7.
Int J Mol Sci ; 25(16)2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39201764

ABSTRACT

Fish retinal ganglion cells (RGCs) can regenerate after optic nerve lesions (ONLs). We previously reported that heat shock factor 1 (HSF1) and Yamanaka factors increased in the zebrafish retina 0.5-24 h after ONLs, and they led to cell survival and the transformation of neuro-stem cells. We also showed that retinoic acid (RA) signaling and transglutaminase 2 (TG2) were activated in the fish retina, performing neurite outgrowth 5-30 days after ONLs. In this study, we found that RA signaling and TG2 increased within 0.5 h in the zebrafish retina after ONLs. We examined their interaction with the TG2-specific morpholino and inhibitor due to the significantly close initiation time of TG2 and HSF1. The inhibition of TG2 led to the complete suppression of HSF1 expression. Furthermore, the results of a ChIP assay with an anti-TG2 antibody evidenced significant anti-TG2 immunoprecipitation of HSF1 genome DNA after ONLs. The inhibition of TG2 also suppressed Yamanaka factors' gene expression. This rapid increase in TG2 expression occurred 30 min after the ONLs, and RA signaling occurred 15 min before this change. The present study demonstrates that TG2 regulates Yamanaka factors via HSF1 signals in the acute phase of fish optic nerve regeneration.


Subject(s)
Heat Shock Transcription Factors , Nerve Regeneration , Optic Nerve , Protein Glutamine gamma Glutamyltransferase 2 , Transglutaminases , Zebrafish , Animals , Zebrafish/genetics , Protein Glutamine gamma Glutamyltransferase 2/metabolism , Transglutaminases/genetics , Transglutaminases/metabolism , Nerve Regeneration/genetics , Optic Nerve/metabolism , Heat Shock Transcription Factors/metabolism , Heat Shock Transcription Factors/genetics , Tretinoin/pharmacology , Tretinoin/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , GTP-Binding Proteins/metabolism , GTP-Binding Proteins/genetics , Retinal Ganglion Cells/metabolism , Gene Expression Regulation/drug effects , Optic Nerve Injuries/metabolism , Optic Nerve Injuries/genetics , Signal Transduction
8.
J Mol Histol ; 55(5): 741-752, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39068616

ABSTRACT

Facial nerve is an integral part of peripheral nerve. Schwann cells are important microglia involved in the repair and regulation of facial nerve injury. LncRNA growth arrest­specific transcript 5 (GAS5) is involved in the behavioral regulation of Schwann cell and the regeneration of peripheral nervous system. However, there is little research about the effect of GAS5 on the repair of facial nerve injury (FNI) by regulating Schwann cells. This study aimed to investigate the role of GAS5 in Schwann cell function and FNI repair, focusing on the miR-138-5p/CXCL12 axis. Hematoxylin and eosin staining, Luxol fast blue staining, transmission electron microscope, and immunofluorescence (IF) experiments were used to verify the effect of GAS5 on FNI rats. Reverse transcription real-time polymerase chain reaction was performed to detect GAS5, miR-138-5p, and C-X-C motif chemokine ligand 12 (CXCL12) mRNA expression. IF staining was used to detect the inflorescence of S100 calcium binding protein B (S100ß), SRY-box transcription factor 10 (SOX10), and tubulin beta 3 class III (ß-Tubulin III). Glial fibrillary acidic protein (GFAP), nerve growth factor receptor (NGFR), S100ß, brain derived neurotrophic factor (BDNF), ciliary neurotrophic factor (CNTF), and CXCL12 proteins were detected using western blot. The 5-bromo-2'-deoxyuridine staining, Transwell, and flow cytometry assays were conducted to detect Schwann cell function. Dual-luciferase, RNA immunoprecipitation, and RNA pulldown assay were used to identify the interaction among GAS5, miR-138-5p, and CXCL12. Results found that GAS5 was downregulated in facial nerve tissues of FNI rats. Overexpressed GAS5 decreased facial grading, inhibited demyelination, and promoted proliferation, migration, and suppressed apoptosis of Schwann cells. Mechanistically, GAS5 was a sponge of miR-138-5p and positively regulated CXCL12 expression. GAS5 inhibition repressed CXCL12 expression and decreased cell proliferation and migration, increased apoptosis rate of Schwann cells by sponging miR-138-5p. In conclusion, overexpression of GAS5 accelerates facial nerve repair in FNI rats by regulating miR-138-5p/CXCL12 axis.


Subject(s)
Chemokine CXCL12 , Facial Nerve Injuries , MicroRNAs , RNA, Long Noncoding , Schwann Cells , Animals , Male , Rats , Cell Proliferation , Chemokine CXCL12/metabolism , Chemokine CXCL12/genetics , Facial Nerve Injuries/metabolism , Facial Nerve Injuries/genetics , Facial Nerve Injuries/pathology , Gene Expression Regulation , MicroRNAs/genetics , MicroRNAs/metabolism , Nerve Regeneration/genetics , Nerve Regeneration/physiology , Rats, Sprague-Dawley , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Schwann Cells/metabolism , Signal Transduction
9.
J Neurosci ; 44(35)2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39054068

ABSTRACT

TFEB and TFE3 (TFEB/3), key regulators of lysosomal biogenesis and autophagy, play diverse roles depending on cell type. This study highlights a hitherto unrecognized role of TFEB/3 crucial for peripheral nerve repair. Specifically, they promote the generation of progenitor-like repair Schwann cells after axonal injury. In Schwann cell-specific TFEB/3 double knock-out mice of either sex, the TFEB/3 loss disrupts the transcriptomic reprogramming that is essential for the formation of repair Schwann cells. Consequently, mutant mice fail to populate the injured nerve with repair Schwann cells and exhibit defects in axon regrowth, target reinnervation, and functional recovery. TFEB/3 deficiency inhibits the expression of injury-responsive repair Schwann cell genes, despite the continued expression of c-jun, a previously identified regulator of repair Schwann cell function. TFEB/3 binding motifs are enriched in the enhancer regions of injury-responsive genes, suggesting their role in repair gene activation. Autophagy-dependent myelin breakdown is not impaired despite TFEB/3 deficiency. These findings underscore a unique role of TFEB/3 in adult Schwann cells that is required for proper peripheral nerve regeneration.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Mice, Knockout , Nerve Regeneration , Peripheral Nerve Injuries , Schwann Cells , Schwann Cells/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Mice , Peripheral Nerve Injuries/metabolism , Nerve Regeneration/physiology , Nerve Regeneration/genetics , Male , Female , Autophagy/physiology , Mice, Inbred C57BL , Sciatic Nerve/injuries
10.
Cell Rep ; 43(7): 114514, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39002126

ABSTRACT

The regenerative potential of injured axons displays considerable heterogeneity. However, the molecular mechanisms underlying the heterogeneity have not been fully elucidated. Here, we establish a method that can separate spinal motor neurons (spMNs) with low and high regenerative capacities and identify a set of transcripts revealing differential expression between two groups of neurons. Interestingly, oligodendrocyte transcription factor 1 (Olig1), which regulates the differentiation of various neuronal progenitors, exhibits recurrent expression in spMNs with enhanced regenerative capabilities. Furthermore, overexpression of Olig1 (Olig1 OE) facilitates axonal regeneration in various models, and down-regulation or deletion of Olig1 exhibits an opposite effect. By analyzing the overlapped differentially expressed genes after expressing individual Olig factor and functional validation, we find that the role of Olig1 is at least partially through the neurite extension factor 1 (Nrsn1). We therefore identify Olig1 as an intrinsic factor that promotes regenerative capacity of injured axons.


Subject(s)
Axons , Basic Helix-Loop-Helix Transcription Factors , Gene Expression Profiling , Nerve Regeneration , Animals , Axons/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Nerve Regeneration/genetics , Nerve Regeneration/physiology , Mice , Motor Neurons/metabolism , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Mice, Inbred C57BL , Transcriptome/genetics
11.
PLoS Genet ; 20(7): e1011367, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39058749

ABSTRACT

The pathway for axon regeneration in Caenorhabditis elegans is activated by SVH-1, a growth factor belonging to the HGF/plasminogen family. SVH-1 is a dual-function factor that acts as an HGF-like growth factor to promote axon regeneration and as a protease to regulate early development. It is important to understand how SVH-1 is converted from a protease to a growth factor for axon regeneration. In this study, we demonstrate that cytidine deaminase (CDD) SVH-17/CDD-2 plays a role in the functional conversion of SVH-1. We find that the codon exchange of His-755 to Tyr in the Asp-His-Ser catalytic triad of SVH-1 can suppress the cdd-2 defect in axon regeneration. Furthermore, the stem hairpin structure around the His-755 site in svh-1 mRNA is required for the activation of axon regeneration by SVH-1. These results suggest that CDD-2 promotes axon regeneration by transforming the function of SVH-1 from a protease to a growth factor through modification of svh-1 mRNA.


Subject(s)
Axons , Caenorhabditis elegans Proteins , Caenorhabditis elegans , Cytidine Deaminase , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Axons/metabolism , Axons/physiology , Cytidine Deaminase/metabolism , Cytidine Deaminase/genetics , Nerve Regeneration/genetics , Nerve Regeneration/physiology , Hepatocyte Growth Factor/metabolism , Hepatocyte Growth Factor/genetics , Regeneration/genetics
12.
Development ; 151(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39007397

ABSTRACT

Many genes are known to regulate retinal regeneration after widespread tissue damage. Conversely, genes controlling regeneration after limited cell loss, as per degenerative diseases, are undefined. As stem/progenitor cell responses scale to injury levels, understanding how the extent and specificity of cell loss impact regenerative processes is important. Here, transgenic zebrafish enabling selective retinal ganglion cell (RGC) ablation were used to identify genes that regulate RGC regeneration. A single cell multiomics-informed screen of 100 genes identified seven knockouts that inhibited and 11 that promoted RGC regeneration. Surprisingly, 35 out of 36 genes known and/or implicated as being required for regeneration after widespread retinal damage were not required for RGC regeneration. The loss of seven even enhanced regeneration kinetics, including the proneural factors neurog1, olig2 and ascl1a. Mechanistic analyses revealed that ascl1a disruption increased the propensity of progenitor cells to produce RGCs, i.e. increased 'fate bias'. These data demonstrate plasticity in the mechanism through which Müller glia convert to a stem-like state and context specificity in how genes function during regeneration. Increased understanding of how the regeneration of disease-relevant cell types is specifically controlled will support the development of disease-tailored regenerative therapeutics.


Subject(s)
Animals, Genetically Modified , Retinal Ganglion Cells , Zebrafish Proteins , Zebrafish , Animals , Zebrafish/genetics , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/physiology , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Nerve Regeneration/genetics , Nerve Regeneration/physiology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , CRISPR-Cas Systems/genetics , Regeneration/genetics , Regeneration/physiology , Retina/metabolism , Retina/cytology , Stem Cells/metabolism , Stem Cells/cytology , Transcription Factors
13.
Gen Comp Endocrinol ; 356: 114579, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38964422

ABSTRACT

The Krüppel-like factors (KLFs) have emerged as important transcriptional regulators of various cellular processes, including neural development. Some of them have been described as intrinsic factors involved in axon regeneration in the central nervous system (CNS) of vertebrates. Zebrafish are known for their ability to regenerate several tissues in adulthood, including the CNS, a capability lost during vertebrate evolution and absent in adult mammals. The role that KLFs could play in this differential ability remains unknown. Therefore, in this study, we analyzed the endogenous response of certain KLFs implicated in axon regeneration (KLFs 6, 7, 9, and 13) during retina development and after axon injury. The results showed that the expression of Klfs 6, 7, and 13 decreases in the developing retina of mice but not in zebrafish, while the mRNA levels of Klf9 strongly increase in both species. The response to injury was further analyzed using optic nerve crush (ONC) as a model of lesion. Our analysis during the acute phase (hours) demonstrated an induction of Klfs 6 and 7 expression exclusively in the zebrafish retina, while Klfs 9 and 13 mRNA levels increased in both species. Further analysis of the chronic response (days) showed that mRNA levels of Klf6 transiently increase in the retinas of both zebrafish and mice, whereas those of Klf7 decrease later after optic nerve injury. In addition, the analysis revealed that the expression of Klf9 decreases, while that of Klf13 increases in the retinas of zebrafish in response to optic nerve injury but remains unaltered in mice. Altogether, these findings support the hypothesis that KLFs may play a role in the differential axon regeneration abilities exhibited by fish and mice.


Subject(s)
Kruppel-Like Transcription Factors , Retina , Zebrafish , Animals , Zebrafish/genetics , Zebrafish/metabolism , Retina/metabolism , Mice , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Optic Nerve Injuries/metabolism , Optic Nerve Injuries/genetics , Nerve Regeneration/physiology , Nerve Regeneration/genetics
14.
Hum Cell ; 37(5): 1394-1404, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38907140

ABSTRACT

The regeneration of peripheral nerves after injury is often slow and impaired, which may be associated with weakened and denervated muscles subsequently leading to atrophy. Adipose-derived stem cells (ADSCs) are often regarded as cell-based therapeutic candidate due to their regenerative potential. The study aims to assess the therapeutic efficacy of gene-modified ADSCs on sciatic nerve injury. We lentivirally transduced ADSCs with shRNA-TWIST1 and transplanted modified cells to rats undergoing sciatic nerve transection and repair. Results showed that TWIST1 knockdown accelerated functional recovery of rats with sciatic nerve injury as faster nerve conduction velocity and higher wire hang scores obtained by rats transplanted with TWIST1-silenced ADSCs than scramble ADSCs. Although the rats experienced degenerated axons and decreased myelin sheath thickness after sciatic nerve injury 8 weeks after operation, those transplanted with TWIST1-silenced ADSCs exhibited more signs of regenerated nerve fibers surrounded by newly formed myelin sheaths than those with scramble ADSCs. The rats transplanted with TWIST1-silenced ADSCs presented increased expressions of neurotrophic factors including neurotrophin-3 (NT-3), brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), and glial cell line-derived neurotrophic factor (GDNF) in the sciatic nerves than those with scramble ADSCs. These results suggest that genetically modifying TWIST1 in ADSCs could facilitate peripheral nerve repair after injury in a more efficient way than that with ADSCs alone.


Subject(s)
Adipose Tissue , Nerve Regeneration , Recovery of Function , Sciatic Nerve , Stem Cell Transplantation , Twist-Related Protein 1 , Animals , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism , Sciatic Nerve/injuries , Nerve Regeneration/genetics , Nerve Regeneration/physiology , Adipose Tissue/cytology , Stem Cell Transplantation/methods , Rats , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Gene Silencing , Stem Cells/metabolism , Stem Cells/cytology , Male , Nerve Growth Factors/metabolism , Nerve Growth Factors/genetics , Rats, Sprague-Dawley , Peripheral Nerve Injuries/therapy , Peripheral Nerve Injuries/genetics , Cells, Cultured , Gene Expression/genetics
15.
Biochim Biophys Acta Mol Cell Res ; 1871(7): 119770, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38897390

ABSTRACT

Microtubule stabilization is critical for axonal growth and regeneration, and many microtubule-associated proteins are involved in this process. In this study, we found that the knockdown of echinoderm microtubule-associated protein-like 1 (EML1) hindered axonal growth in cultured cortical and dorsal root ganglion neurons. We further revealed that EML1 facilitated the acetylation of microtubules and that the impairment of axonal growth due to EML1 inhibition could be restored by treatment with deacetylase inhibitors, suggesting that EML1 affected tubulin acetylation. Moreover, we verified an interaction between EML1 and the alpha-tubulin acetyltransferase 1, which is responsible for the acetylation of alpha-tubulin. We thus proposed that EML1 might regulate microtubule acetylation and stabilization via alpha-tubulin acetyltransferase 1 and then promote axon growth. Finally, we verified that the knockdown of EML1 in vivo also inhibited sciatic nerve regeneration. Our findings revealed a novel effect of EML1 on microtubule acetylation during axonal regeneration.


Subject(s)
Acetyltransferases , Amino Acid Transport System A , Axons , Microtubule-Associated Proteins , Animals , Humans , Mice , Rats , Acetylation , Acetyltransferases/metabolism , Acetyltransferases/genetics , Axons/metabolism , Cells, Cultured , Ganglia, Spinal/metabolism , Ganglia, Spinal/cytology , Microtubule Proteins , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Microtubules/metabolism , Nerve Regeneration/genetics , Sciatic Nerve/metabolism , Tubulin/metabolism , Tubulin/genetics , Amino Acid Transport System A/metabolism
16.
Sci Adv ; 10(26): eadm8454, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38941462

ABSTRACT

The formation of vascular niche is pivotal during the early stage of peripheral nerve regeneration. Nevertheless, the mechanisms of vascular niche in the regulation of peripheral nerve repair remain unclear. Netrin-1 (NTN1) was found up-regulated in nerve stump after peripheral nerve injury (PNI). Herein, we demonstrated that NTN1-high endothelial cells (NTN1+ECs) were the critical component of vascular niche, fostering angiogenesis, axon regeneration, and repair-related phenotypes. We also found that NTN1+EC-derived exosomes (NTN1 EC-EXO) were involved in the formation of vascular niche as a critical role. Multi-omics analysis further verified that NTN1 EC-EXO carried a low-level expression of let7a-5p and activated key pathways associated with niche formation including focal adhesion, axon guidance, phosphatidylinositol 3-kinase-AKT, and mammalian target of rapamycin signaling pathway. Together, our study suggested that the construction of a pre-regenerative niche induced by NTN1 EC-EXO could establish a beneficial microenvironment for nerve repair and facilitate functional recovery after PNI.


Subject(s)
Endothelial Cells , Exosomes , Nerve Regeneration , Netrin-1 , Peripheral Nerve Injuries , Netrin-1/metabolism , Netrin-1/genetics , Exosomes/metabolism , Nerve Regeneration/genetics , Animals , Endothelial Cells/metabolism , Peripheral Nerve Injuries/metabolism , Peripheral Nerve Injuries/therapy , Peripheral Nerve Injuries/pathology , Mice , Neovascularization, Physiologic , Signal Transduction , Humans , Peripheral Nerves/metabolism
17.
Neurosci Lett ; 833: 137813, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38723761

ABSTRACT

A significant public health burden is peripheral nerve damage (PNI), which is frequently brought on by trauma. Macrophages were essential to the effective regeneration of nerves and restoration of function. It is still not entirely understood how macrophages and Schwann cells interact after damage during remyelination. Here, we established an inflammatory model in bone marrow-derived macrophages (BMDMs) and a rat sciatic nerve damage model to investigate the possible relationship between lipopolysaccharides (LPS)-induced exosomes derived from Schwann cells (LPS SCs-Exos) and peripheral nerve repair. The pro-inflammatory macrophage was changed into a pro-regeneration macrophage by LPS SC-Exos. Notably, it was discovered that SC-Exos had a substantial enrichment of OTULIN. OTULIN was a key mediator in the regulatory effects of LPS SC-Exos by deubiquitinating ERBB2 and preventing its degradation. The local injection of SC-Exos into the nerve damage site led in a faster functional recovery, axon regeneration and remyelination, and an increased M2 macrophage polarization, whereas OTULIN knockdown reversed these effects in vivo. Our results indicate that LPS SC-Exos may offer a therapeutic avenue for peripheral nerve regeneration by promoting macrophage polarization toward an M2 phenotype through the shuttling of OTULIN and deubiquitination of ERBB2. SIGNIFICANCE STATEMENT: OTULIN protein from SC-Exos mediated the macrophages polarization and axonal growth in BMDMs through promoting ubiquitination of ERBB2 and triggering the degradation of ERBB2. The findings offered prospective therapeutic hints for PNI therapy approaches that target axonal regrowth.


Subject(s)
Endopeptidases , Exosomes , Macrophages , Nerve Regeneration , Peripheral Nerve Injuries , Schwann Cells , Animals , Rats , Exosomes/metabolism , Lipopolysaccharides , Macrophages/metabolism , Nerve Regeneration/genetics , Nerve Regeneration/physiology , Peripheral Nerve Injuries/genetics , Peripheral Nerve Injuries/metabolism , Rats, Sprague-Dawley , Receptor, ErbB-2/metabolism , Schwann Cells/metabolism , Sciatic Nerve/injuries , Sciatic Nerve/metabolism , Ubiquitination , Endopeptidases/genetics , Endopeptidases/metabolism
18.
Neurosci Lett ; 833: 137832, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38796094

ABSTRACT

Axonal regeneration is restricted in adults and causes irreversible motor dysfunction following spinal cord injury (SCI). In contrast, neonates have prominent regenerative potential and can restore their neural function. Although the distinct cellular responses in neonates have been studied, how they contribute to neural recovery remains unclear. To assess whether the secreted molecules in neonatal SCI can enhance neural regeneration, we re-analyzed the previously performed single-nucleus RNA-seq (snRNA-seq) and focused on Asporin and Cd109, the highly expressed genes in the injured neonatal spinal cord. In the present study, we showed that both these molecules were expressed in the injured spinal cords of adults and neonates. We treated the cortical neurons with recombinant Asporin or CD109 to observe their direct effects on neurons in vitro. We demonstrated that these molecules enhance neurite outgrowth in neurons. However, these molecules did not enhance re-growth of severed axons. Our results suggest that Asporin and CD109 influence neurites at the lesion site, rather than promoting axon regeneration, to restore neural function in neonates after SCI.


Subject(s)
Axons , Extracellular Matrix Proteins , Nerve Regeneration , Spinal Cord Injuries , Animals , Animals, Newborn , Antigens, CD/metabolism , Axons/metabolism , Cells, Cultured , Nerve Regeneration/genetics , Nerve Regeneration/physiology , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Neurites/metabolism , Neurites/drug effects , Neuronal Outgrowth/physiology , Neurons/metabolism , Spinal Cord/metabolism , Spinal Cord Injuries/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Mice
19.
Cell Rep Med ; 5(5): 101554, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38729157

ABSTRACT

The axons of retinal ganglion cells (RGCs) form the optic nerve, transmitting visual information from the eye to the brain. Damage or loss of RGCs and their axons is the leading cause of visual functional defects in traumatic injury and degenerative diseases such as glaucoma. However, there are no effective clinical treatments for nerve damage in these neurodegenerative diseases. Here, we report that LIM homeodomain transcription factor Lhx2 promotes RGC survival and axon regeneration in multiple animal models mimicking glaucoma disease. Furthermore, following N-methyl-D-aspartate (NMDA)-induced excitotoxicity damage of RGCs, Lhx2 mitigates the loss of visual signal transduction. Mechanistic analysis revealed that overexpression of Lhx2 supports axon regeneration by systematically regulating the transcription of regeneration-related genes and inhibiting transcription of Semaphorin 3C (Sema3C). Collectively, our studies identify a critical role of Lhx2 in promoting RGC survival and axon regeneration, providing a promising neural repair strategy for glaucomatous neurodegeneration.


Subject(s)
Axons , Disease Models, Animal , Glaucoma , LIM-Homeodomain Proteins , Nerve Regeneration , Retinal Ganglion Cells , Transcription Factors , Animals , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , LIM-Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/genetics , Glaucoma/genetics , Glaucoma/pathology , Glaucoma/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Axons/metabolism , Axons/pathology , Mice , Nerve Regeneration/genetics , Nerve Regeneration/physiology , Mice, Inbred C57BL , Cell Survival/genetics , Semaphorins/metabolism , Semaphorins/genetics , N-Methylaspartate/metabolism
20.
Cell Commun Signal ; 22(1): 240, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38664711

ABSTRACT

BACKGROUND: The repair of peripheral nerve injury poses a clinical challenge, necessitating further investigation into novel therapeutic approaches. In recent years, bone marrow mesenchymal stromal cell (MSC)-derived mitochondrial transfer has emerged as a promising therapy for cellular injury, with reported applications in central nerve injury. However, its potential therapeutic effect on peripheral nerve injury remains unclear. METHODS: We established a mouse sciatic nerve crush injury model. Mitochondria extracted from MSCs were intraneurally injected into the injured sciatic nerves. Axonal regeneration was observed through whole-mount nerve imaging. The dorsal root ganglions (DRGs) corresponding to the injured nerve were harvested to test the gene expression, reactive oxygen species (ROS) levels, as well as the degree and location of DNA double strand breaks (DSBs). RESULTS: The in vivo experiments showed that the mitochondrial injection therapy effectively promoted axon regeneration in injured sciatic nerves. Four days after injection of fluorescently labeled mitochondria into the injured nerves, fluorescently labeled mitochondria were detected in the corresponding DRGs. RNA-seq and qPCR results showed that the mitochondrial injection therapy enhanced the expression of Atf3 and other regeneration-associated genes in DRG neurons. Knocking down of Atf3 in DRGs by siRNA could diminish the therapeutic effect of mitochondrial injection. Subsequent experiments showed that mitochondrial injection therapy could increase the levels of ROS and DSBs in injury-associated DRG neurons, with this increase being correlated with Atf3 expression. ChIP and Co-IP experiments revealed an elevation of DSB levels within the transcription initiation region of the Atf3 gene following mitochondrial injection therapy, while also demonstrating a spatial proximity between mitochondria-induced DSBs and CTCF binding sites. CONCLUSION: These findings suggest that MSC-derived mitochondria injected into the injured nerves can be retrogradely transferred to DRG neuron somas via axoplasmic transport, and increase the DSBs at the transcription initiation regions of the Atf3 gene through ROS accumulation, which rapidly release the CTCF-mediated topological constraints on chromatin interactions. This process may enhance spatial interactions between the Atf3 promoter and enhancer, ultimately promoting Atf3 expression. The up-regulation of Atf3 induced by mitochondria further promotes the expression of downstream regeneration-associated genes and facilitates axon regeneration.


Subject(s)
Activating Transcription Factor 3 , Axons , DNA Breaks, Double-Stranded , Ganglia, Spinal , Mesenchymal Stem Cells , Mitochondria , Nerve Regeneration , Reactive Oxygen Species , Sciatic Nerve , Up-Regulation , Animals , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Mitochondria/metabolism , Mitochondria/genetics , Reactive Oxygen Species/metabolism , Axons/metabolism , Nerve Regeneration/genetics , Up-Regulation/genetics , Mice , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Sciatic Nerve/injuries , Sciatic Nerve/pathology , Ganglia, Spinal/metabolism , Mice, Inbred C57BL , Male
SELECTION OF CITATIONS
SEARCH DETAIL